Combination Chemotherapy May Significantly Improve Treatment for Glioblastoma

Summary: Researchers report the addition of an FDA approved chemotherapy drug called hydroxyurea to temozolomide helps to increase survival rates in animal models of glioblastoma.

Source: Mass General.

A diagnosis of the brain cancer glioblastoma carries a dismal prognosis, with most patients dying within five years. Now a team led by Massachusetts General Hospital (MGH) investigators has found that adding the chemotherapy drug hydroxyurea to the current chemotherapy protocol for glioblastoma significantly increased survival in animal models. The team’s report has been published in the journal Neuro-Oncology.

“Through drug screening aimed at repurposing old drugs, we demonstrated that the FDA-approved drug hydroxyurea synergizes with temozolomide in glioblastoma cells from newly diagnosed and recurrent tumors, including those that were resistant to treatment” says Bakhos Tannous, PhD, of the Neuro-Oncology Division in the MGH Department of Neurology, senior author of the report. “The combination led to a significant increase in survival in different mouse models, in some of which 40 to 60 percent of animals became tumor-free.”

The addition of temozolomide (TMZ) to surgical and radiation treatment has been the major improvement in glioblastoma treatment over the past 20 years, raising two-year survival from 11 percent to 27 percent. But 90 percent of patients receiving that combination still die within five years, representing what the authors call “a colossal failure.” The major factor leading to that failure has been the development of resistance to TMZ, which acts by inhibiting the synthesis of proteins within cancer cells.

Much of that resistance depends on the action of an enzyme called MGMT, which repairs the DNA alteration induced by TMZ. It is known that whether or not the MGMT promoter is methylated – a DNA modification that suppresses the expression of a gene – can predict whether a glioblastoma tumor will respond to TMZ. A change in methylation status that activates MGMT may be behind the initial resistance of tumors to TMZ, but other resistance mechanisms are also being investigated.

In their search for ways to overcome resistance to TMZ, the MGH-led team pursued a strategy of screening drugs already approved for the treatment of several types of cancer to see if they could improve glioblastoma outcomes. Tannous explains that the development of new drugs is a long, difficult and expensive process that often fails due to unexpected side effects in patients. But much is already known about the pharmacology, mechanisms of action and potential toxicities of existing drugs. “The advantage of ‘teaching old drugs new tricks’ is that, compared with typical drug discovery, clinical use could have a much faster transition into the clinic,” he says.

The team screened a library of 21 anti-cancer drugs – both newer targeted agents and commonly used traditional chemotherapy drugs – against 18 cultured glioblastoma cell lines, including stem-like cells from newly diagnosed and recurrent tumors that were both sensitive and resistant to TMZ treatment. While several agents improved the response of some TMZ-resistant cultures, only hydroxyurea sensitized practically all cell lines – including several that were previously resistant – to TMZ treatment.

brain slice
In their search for ways to overcome resistance to TMZ, the MGH-led team pursued a strategy of screening drugs already approved for the treatment of several types of cancer to see if they could improve glioblastoma outcomes. NeuroscienceNews.com image is in the public domain.

With these promising results, the team investigated that combination in several mouse models of glioblastoma, including tumors with different MGMT status. While either drug alone had only a moderate effect on tumor growth and survival, the combination of hydroxyurea and TMZ significantly improved both. Overall, around half of the animals receiving the combination treatment remained tumor-free at the end of the study period, and in those with a specific subtype of tumor, 60 percent were essentially cured.

“The fact that the combination of hydroxyurea and TMZ was effective in all of the cellular and animal models and not selective for a specific type of tumor is very exciting, since it suggests that the combination would be effective in all glioblastoma patients, not only those with inactive MGMT,” says Tannous, who is an associate professor of Neurology at Harvard Medical School. “Hydroxyurea has been used for decades to treat conditions such as sickle cell disease and some types of cancer, so bringing this approach into the clinic should be straightforward. We are currently preparing to test this combination in a phase 1 clinical trial, which we hope to start very soon.

About this neuroscience research article

The co-senior authors of the Neuro-Oncology paper are Thomas Wurdinger, PhD, MGH Neuro-Oncology Division and VU University Medical Center, Amsterdam; and Tracy Batchelor, MD, Pappas Center for Neuro-Oncology, MGH Cancer Center. The lead author is Jian Teng, PhD, MGH Neuro-Oncology. Additional co-authors are Seyedali Hejazi, Litia Carvalho, PhD, and Marco Barazas, MGH Neuro-Oncology; Hiroaki Wakimoto, MD, PhD, MGH Neurosurgery; Lotte Hiddingh, Mark de Gooijer, David Noske, MD, and Pieter Wesseling, MD, VU University Medical Center; Marie Tannous, Notre Dame University-Louaize, Lebanon; and Andrew S. Chi, MD, PhD, New York University/Langone Medical Center.

Funding: The study was supported by National Institutes of Health grants R01 NS064983, 1R01 NS095647, P30 NS04776 and R01 CA166077 and grants from Stichting Stophersentumoren of the Netherlands. The MGH has filed a patent application for the use of hydroxyurea and similar drugs in combination with TMZ to treat glioblastoma.

Source: Katie Marquedant – Mass General
Publisher: Organized by NeuroscienceNews.com.
Image Source: NeuroscienceNews.com image is in the public domain.
Original Research: Abstract in Neuro-Oncology
doi: 10.1093/neuonc/nox198

Cite This NeuroscienceNews.com Article

[cbtabs][cbtab title=”MLA”]Mass General “Combination Chemotherapy May Significantly Improve Treatment for Glioblastoma.” NeuroscienceNews. NeuroscienceNews, 22 January 2018.
<https://neurosciencenews.com/chemo-combination-gbm-8341/>.[/cbtab][cbtab title=”APA”]Mass General (2018, January 22). Combination Chemotherapy May Significantly Improve Treatment for Glioblastoma. NeuroscienceNews. Retrieved January 22, 2018 from https://neurosciencenews.com/chemo-combination-gbm-8341/[/cbtab][cbtab title=”Chicago”]Mass General “Combination Chemotherapy May Significantly Improve Treatment for Glioblastoma.” https://neurosciencenews.com/chemo-combination-gbm-8341/ (accessed January 22, 2018).[/cbtab][/cbtabs]


Abstract

Recycling drug screen repurposes hydroxyurea as a sensitizer of glioblastomas to temozolomide targeting de novo DNA synthesis, irrespective of molecular subtype

Background
Glioblastoma (GBM) is the most common and most aggressive primary malignant brain tumor. Standard-of-care treatment involves maximal surgical resection of the tumor followed by radiation and chemotherapy (temozolomide [TMZ]). The 5-year survival rate of patients with GBM is <10%, a colossal failure that has been partially attributed to intrinsic and/or acquired resistance to TMZ through O6-methylguanine DNA methyltransferase (MGMT) promoter methylation status in the tumor.

Methods
A drug screening aimed at evaluating the potential recycling and repurposing of known drugs was conducted in TMZ-resistant GBM cell lines and primary cultures of newly diagnosed GBM with different MGMT promoter methylation status, phenotypic/genotypic background and subtype, and validated with sphere formation, cell migration assays, and quantitative invasive orthotopic in vivo models.

Results
We identified hydroxyurea (HU) to synergize with TMZ in GBM cells in culture and in vivo, irrespective of MGMT promoter methylation status, subtype, and/or stemness. HU acts specifically on the S-phase of the cell cycle by inhibiting the M2 unit of enzyme ribonucleotide reductase. Knockdown of this enzyme using RNA interference and other known chemical inhibitors exerted a similar effect to HU in combination with TMZ both in culture and in vivo.

Conclusions
We demonstrate preclinical efficacy of repurposing hydroxyurea in combination with TMZ for adjuvant GBM therapy. This combination benefit is of direct clinical interest given the extensive use of TMZ and the associated problems with TMZ-related resistance and treatment failure.

Feel free to share this Neuroscience News.
Join our Newsletter
I agree to have my personal information transferred to AWeber for Neuroscience Newsletter ( more information )
Sign up to receive our recent neuroscience headlines and summaries sent to your email once a day, totally free.
We hate spam and only use your email to contact you about newsletters. You can cancel your subscription any time.